Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 281
Filter
Add more filters

Publication year range
1.
Nano Lett ; 24(37): 11520-11528, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39234992

ABSTRACT

Small-molecule biomarkers are ubiquitous in biological fluids with pathological implications, but major challenges persist in their quantitative analysis directly in complex clinical samples. Herein, a molecular-sieving label-free surface-enhanced Raman spectroscopy (SERS) biosensor is reported for selective quantitative analysis of trace small-molecule trimetazidine (TMZ) in clinical samples. Our biosensor is fabricated by decorating a superhydrophobic monolayer of microporous metal-organic frameworks (MOF) shell-coated Au nanostar nanoparticles on a silicon substrate. The design strategy principally combines the hydrophobic surface-enabled physical confinement and preconcentration, MOF-assisted molecular enrichment and sieving of small molecules, and sensitive SERS detection. Our biosensor utilizes such a "molecular confinement-and-sieving" strategy to achieve a five orders-of-magnitude dynamic detection range and a limit of detection of ≈0.5 nM for TMZ detection in either urine or whole blood. We further demonstrate the applicability of our biosensing platform for longitudinal label-free SERS detection of the TMZ level directly in clinical samples in a mouse model.


Subject(s)
Biosensing Techniques , Gold , Metal Nanoparticles , Metal-Organic Frameworks , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , Animals , Mice , Gold/chemistry , Biosensing Techniques/methods , Metal Nanoparticles/chemistry , Humans , Metal-Organic Frameworks/chemistry , Biomarkers/urine , Biomarkers/analysis , Surface Properties , Limit of Detection
2.
FASEB J ; 37(2): e22768, 2023 02.
Article in English | MEDLINE | ID: mdl-36624703

ABSTRACT

Platelet mitochondria possess remarkable plasticity for oxidation of energy substrates, where metabolic dependency on glucose or fatty acids is higher than glutamine. Since platelets metabolize nearly the entire pool of glucose to lactate rather than fluxing through mitochondrial tricarboxylic acid cycle, we posit that majority of mitochondrial ATP, which is essential for platelet granule secretion and thrombus formation, is sourced from oxidation of fatty acids. We performed a comprehensive analysis of bioenergetics and function of stimulated platelets in the presence of etomoxir, trimetazidine and oxfenicine, three pharmacologically distinct inhibitors of ß-oxidation. Each of them significantly impaired oxidative phosphorylation in unstimulated as well as thrombin-stimulated platelets leading to a small but consistent drop in ATP level in activated cells due to a lack of compensation from glycolytic ATP. Trimetazidine and oxfenicine attenuated platelet aggregation, P-selectin externalization and integrin αIIb ß3 activation. Both etomoxir and trimetazidine impeded agonist-induced dense granule release and platelet thrombus formation on collagen under arterial shear. The effect of inhibitors on platelet aggregation and dense granule release was dose- and incubation time- dependent with significant inhibition at higher doses and prolonged incubation times. Neither of the inhibitors could protect mice from collagen-epinephrine-induced pulmonary embolism or prolong mouse tail bleeding times. However, mice pre-administered with etomoxir, trimetazidine and oxfenicine were protected from ferric chloride-induced mesenteric thrombosis. In conclusion, ß-oxidation of fatty acids sustains ATP level in stimulated platelets and is therefore essential for energy-intensive agonist-induced platelet responses. Thus, fatty acid oxidation may constitute an attractive therapeutic target for novel antiplatelet agents.


Subject(s)
Thrombosis , Trimetazidine , Mice , Animals , Fatty Acids/metabolism , Trimetazidine/adverse effects , Trimetazidine/metabolism , Blood Platelets/metabolism , Platelet Activation , Platelet Aggregation , Thrombosis/chemically induced , Thrombosis/prevention & control , Thrombosis/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/adverse effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Oxidative Phosphorylation , Collagen/metabolism , Adenosine Triphosphate/metabolism , Glucose/metabolism
3.
Pharmacol Res ; 206: 107281, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942341

ABSTRACT

Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.


Subject(s)
Cardiovascular Diseases , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Animals , Apoptosis/drug effects , Signal Transduction , Cardiovascular Agents/therapeutic use , Cardiovascular Agents/pharmacology
4.
Int J Mol Sci ; 25(3)2024 Jan 28.
Article in English | MEDLINE | ID: mdl-38338885

ABSTRACT

Trimetazidine (TMZ), used for treating stable angina pectoris, has garnered attention in the realm of sports due to its potential performance-enhancing properties, and the World Anti-Doping Agency (WADA) has classified TMZ on the S4 list of prohibited substances since 2014. The purpose of this narrative mini-review is to emphasize the biochemical aspects underlying the abusive use of TMZ among athletes as a metabolic modulator of cardiac energy metabolism. The myocardium's ability to adapt its energy substrate utilization between glucose and fatty acids is crucial for maintaining cardiac function under various conditions, such as rest, moderate exercise, and intense effort. TMZ acts as a partial inhibitor of fatty acid oxidation by inhibiting 3-ketoacyl-CoA thiolase (KAT), shifting energy production from long-chain fatty acids to glucose, reducing oxygen consumption, improving cardiac function, and enhancing exercise capacity. Furthermore, TMZ modulates pyruvate dehydrogenase (PDH) activity, promoting glucose oxidation while lowering lactate production, and ultimately stabilizing myocardial function. TMZs role in reducing oxidative stress is notable, as it activates antioxidant enzymes like glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD). In conclusion, TMZs biochemical mechanisms make it an attractive but controversial option for athletes seeking a competitive edge.


Subject(s)
Trimetazidine , Humans , Trimetazidine/pharmacology , Trimetazidine/therapeutic use , Vasodilator Agents/pharmacology , Fatty Acids/metabolism , Glucose/metabolism , Athletes
5.
Int J Mol Sci ; 25(6)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38542223

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is considered the prototype of motor neuron disease, characterized by motor neuron loss and muscle waste. A well-established pathogenic hallmark of ALS is mitochondrial failure, leading to bioenergetic deficits. So far, pharmacological interventions for the disease have proven ineffective. Trimetazidine (TMZ) is described as a metabolic modulator acting on different cellular pathways. Its efficacy in enhancing muscular and cardiovascular performance has been widely described, although its molecular target remains elusive. We addressed the molecular mechanisms underlying TMZ action on neuronal experimental paradigms. To this aim, we treated murine SOD1G93A-model-derived primary cultures of cortical and spinal enriched motor neurons, as well as a murine motor-neuron-like cell line overexpressing SOD1G93A, with TMZ. We first characterized the bioenergetic profile of the cell cultures, demonstrating significant mitochondrial dysfunction that is reversed by acute TMZ treatments. We then investigated the effect of TMZ in promoting autophagy processes and its impact on mitochondrial morphology. Finally, we demonstrated the effectiveness of TMZ in terms of the mitochondrial functionality of ALS-rpatient-derived peripheral blood mononuclear cells (PBMCs). In summary, our results emphasize the concept that targeting mitochondrial dysfunction may represent an effective therapeutic strategy for ALS. The findings demonstrate that TMZ enhances mitochondrial performance in motor neuron cells by activating autophagy processes, particularly mitophagy. Although further investigations are needed to elucidate the precise molecular pathways involved, these results hold critical implications for the development of more effective and specific derivatives of TMZ for ALS treatment.


Subject(s)
Amyotrophic Lateral Sclerosis , Mitochondrial Diseases , Trimetazidine , Mice , Animals , Humans , Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase-1/metabolism , Trimetazidine/pharmacology , Trimetazidine/therapeutic use , Mice, Transgenic , Leukocytes, Mononuclear/metabolism , Superoxide Dismutase/metabolism , Autophagy , Disease Models, Animal
6.
J Pak Med Assoc ; 74(6 (Supple-6)): S28-S33, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39018136

ABSTRACT

OBJECTIVE: To examine the impact of trimetazidine on skeletal muscle function in patients suffering from peripheral artery disease. METHODS: The systematic review was conducted from July 20 to November 22, 2022, in line with the Preferred Reporting Items for Systematic Review and Meta-Analysis and comprised search for interventional studies on MEDLINE, ProQuest, Scopus and ScienceDirect databases using key words "peripheral artery disease" and "trimetazidine" or their synonyms. The cut-off date for the search was July 21, 2022. Clinical parameters, including Ankle-Brachial Index, Maximum Walking Distance, Maximum Walking Time and Pain Onset Time, were analysed both narratively and quantitatively whenever possible. RESULTS: Of the 587 studies initially identified, 12(2%) were shortlisted. Of them, 2(16.7%) qualified for detailed analysis, comprising 172 patients with intermittent claudication. There was no significant difference between the examined groups' Ankle-Brachial Index values at baseline and post-intervention (p=0.83). Maximum Walking Distance improvement was significantly higher (p=0.0006) in trimetazidine group compared to control group. Maximum Walking Time MWT and Pain Onset Time were significantly different between control and trimetazidine groups (p<0.05). CONCLUSIONS: Trimetazidine's anti-ischaemic effect in peripheral artery disease patients improved Maximum Walking Distance, while it had no significant influence on Ankle-Brachial Index. Well-designed studies addressing the issue are needed.


Subject(s)
Ankle Brachial Index , Peripheral Arterial Disease , Trimetazidine , Vasodilator Agents , Trimetazidine/therapeutic use , Humans , Peripheral Arterial Disease/drug therapy , Peripheral Arterial Disease/physiopathology , Vasodilator Agents/therapeutic use , Walking/physiology , Intermittent Claudication/drug therapy , Intermittent Claudication/physiopathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/blood supply , Walk Test
7.
Bull Exp Biol Med ; 177(2): 235-237, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39090459

ABSTRACT

We studied the effect of N1-(2,3,4-trimethoxybenzyl)-N2-{2-[(2,3,4-trimethoxybenzyl)amino]ethyl}-1,2-ethanediamine (compound ALM-802) on the physical performance of mice after acute fatigue. The animals' performance was assessed on a treadmill. The criterion for assessing exercise tolerance was the length of the distance passed when running on a treadmill until complete fatigue. To assess the actoprotective activity of compound ALM-802, we used a method of stepwise increase in load with an initial running speed of 42 cm/sec and its subsequent increase by 5 cm/sec every 5 min. The maximum speed of movement of the treadmill belt is 77 cm/sec. Animals that received compound ALM-802 (2 mg/kg intraperitoneally), 1 day after acute fatigue, ran a distance to complete fatigue that exceeded that of control mice by 68% (387.9±60.5 and 230.6±29.6 m, respectively, p=0.023). The reference drug trimetazidine (30 mg/kg, intraperitoneally) did not have a significant effect on the distance traveled. Compound ALM-802 helps restore physical performance, i.e. exhibits significant actoprotective activity.


Subject(s)
Fatigue , Animals , Mice , Male , Fatigue/drug therapy , Exercise Tolerance/drug effects , Physical Conditioning, Animal , Physical Functional Performance , Diamines/chemistry , Diamines/pharmacology
8.
Mol Syst Biol ; 18(9): e10716, 2022 09.
Article in English | MEDLINE | ID: mdl-36094015

ABSTRACT

Quiescent cancer cells are rare nondiving cells with the unique ability to evade chemotherapies and resume cell division after treatment. Despite the associated risk of cancer recurrence, how cells can reversibly switch between rapid proliferation and quiescence remains a long-standing open question. By developing a unique methodology for the cell sorting-free separation of metabolic profiles in cell subpopulations in vitro, we unraveled metabolic characteristics of quiescent cells that are largely invariant to basal differences in cell types and quiescence-inducing stimuli. Consistent with our metabolome-based analysis, we show that impairing mitochondrial fatty acid ß-oxidation (FAO) can induce apoptosis in quiescence-induced cells and hamper their return to proliferation. Our findings suggest that in addition to mediating energy and redox balance, FAO can play a role in preventing the buildup of toxic intermediates during transitioning to quiescence. Uncovering metabolic strategies to enter, maintain, and exit quiescence can reveal fundamental principles in cell plasticity and new potential therapeutic targets beyond cancer.


Subject(s)
Fatty Acids , Metabolomics , Cell Division , Cell Movement , Protein Transport
9.
Bipolar Disord ; 25(8): 661-670, 2023 12.
Article in English | MEDLINE | ID: mdl-36890661

ABSTRACT

OBJECTIVES: The aim of this study was to repurpose a drug for the treatment of bipolar depression. METHODS: A gene expression signature representing the overall transcriptomic effects of a cocktail of drugs widely prescribed to treat bipolar disorder was generated using human neuronal-like (NT2-N) cells. A compound library of 960 approved, off-patent drugs were then screened to identify those drugs that affect transcription most similar to the effects of the bipolar depression drug cocktail. For mechanistic studies, peripheral blood mononuclear cells were obtained from a healthy subject and reprogrammed into induced pluripotent stem cells, which were then differentiated into co-cultured neurons and astrocytes. Efficacy studies were conducted in two animal models of depressive-like behaviours (Flinders Sensitive Line rats and social isolation with chronic restraint stress rats). RESULTS: The screen identified trimetazidine as a potential drug for repurposing. Trimetazidine alters metabolic processes to increase ATP production, which is thought to be deficient in bipolar depression. We showed that trimetazidine increased mitochondrial respiration in cultured human neuronal-like cells. Transcriptomic analysis in induced pluripotent stem cell-derived neuron/astrocyte co-cultures suggested additional mechanisms of action via the focal adhesion and MAPK signalling pathways. In two different rodent models of depressive-like behaviours, trimetazidine exhibited antidepressant-like activity with reduced anhedonia and reduced immobility in the forced swim test. CONCLUSION: Collectively our data support the repurposing of trimetazidine for the treatment of bipolar depression.


Subject(s)
Bipolar Disorder , Trimetazidine , Rats , Humans , Animals , Trimetazidine/pharmacology , Trimetazidine/therapeutic use , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Transcriptome , Drug Repositioning , Leukocytes, Mononuclear , Disease Models, Animal
10.
Cardiovasc Drugs Ther ; 37(3): 497-506, 2023 06.
Article in English | MEDLINE | ID: mdl-34767131

ABSTRACT

PURPOSE: Trimetazidine, a metabolic agent with anti-ischemic effects, was reported to reduce reperfusion injury in animal models. In this randomized double-blind placebo-controlled trial, we investigated the effects of trimetazidine on the reduction of infarction size in patients undergoing revascularization for ST segment elevation myocardial infarction (STEMI). METHODS: Patients with STEMI randomly received trimetazidine (n = 87) or placebo (n = 86) before primary percutaneous coronary intervention (PCI), and subsequently received oral trimetazidine or placebo for 12 months after reperfusion. The predefined primary endpoint was infarction size on cardiac magnetic resonance (CMR) performed at 7 days after primary PCI. The trial was registered on www. CLINICALTRIALS: gov (registration number: NCT02826616). RESULTS: The clinical characteristics of the patients in both groups were well-matched at baseline. At 7 days after primary PCI, the percentage and absolute infarction size in the trimetazidine group were significantly smaller than those in the control group (22% ± 12% [n = 74] vs. 27% ± 13% [n = 74], p = 0.011 and 28 ± 18 g [n = 74] vs. 35 ± 19 g [n = 74], p = 0.022, respectively), and the incidence of myocardial microvascular obstruction (MVO) measured by CMR was significantly reduced in the trimetazidine group (29.7% [22/74] vs. 52.7% [39/74], p = 0.005). The myocardial salvage index (MSI) measured by CMR was significantly higher in the trimetazidine group (48% ± 20% vs. 39% ± 20%, p = 0.008). The incidence of readmission due to aggravated heart failure did not differ significantly between the trimetazidine group and the control group (8.0% vs. 14.0%, p = 0.234). CONCLUSIONS: In patients with STEMI undergoing primary PCI, early trimetazidine before reperfusion reduced myocardial infarction size and MVO, and improved MSI.


Subject(s)
Myocardial Infarction , Percutaneous Coronary Intervention , ST Elevation Myocardial Infarction , Trimetazidine , Humans , ST Elevation Myocardial Infarction/diagnostic imaging , ST Elevation Myocardial Infarction/therapy , Trimetazidine/adverse effects , Percutaneous Coronary Intervention/adverse effects , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/drug therapy , Myocardium/pathology , Treatment Outcome
11.
Cardiovasc Drugs Ther ; 37(5): 955-964, 2023 10.
Article in English | MEDLINE | ID: mdl-35538296

ABSTRACT

BACKGROUND: Previous studies have shown that Trimetazidine (TMZ) improves vascular endothelial function and reduces the inflammatory process progression. However, limited data have been available regarding its effects on myocardial fibrosis following ischemia and causing left ventricular dysfunction. PURPOSE: To investigate the impact of TMZ adjuvant therapy for ischemic cardiomyopathy (ICM) on cardiac fibrosis, vascular endothelial function, inflammation, and myocardial functions. METHODS: This randomized, double-blind controlled clinical trial included 48 patients (aged 59.4 ± 9 years) with ICM who were randomly assigned to two groups: TMZ 35 mg twice daily and placebo in addition to conventional ICM medications. All patients received the tablets for 3 months. Both groups were then compared in terms of connective tissue growth factor (CTGF), endothelin-1 (ET-1), tumor necrosis factor-alpha (TNF-α), and some echocardiographic indices, weekly angina attacks and nitrate consumption before and after treatment. RESULTS: No significant differences between CTGF, ET-1, and TNF-α levels, in addition to some echocardiographic indices, were observed between both groups before treatment. After treatment, the TMZ group had significantly lower ET-1 than the placebo group, with both groups exhibiting a substantial decrease in TNF-α and CTGF. The TMZ group had lower mean ± SD levels for TNF-α and CTGF and showed significant improvement in echocardiographic indices and weekly angina attacks after treatment. CONCLUSION: Adjunctive TMZ therapy for ICM effectively improved vascular endothelial function and reduced inflammation. Furthermore, our exploratory findings may be used to provide new information on the potential effects of TMZ on myocardial fibrosis by downregulating CTGF.


Subject(s)
Cardiomyopathies , Myocardial Ischemia , Trimetazidine , Humans , Trimetazidine/adverse effects , Vasodilator Agents/adverse effects , Tumor Necrosis Factor-alpha , Myocardial Ischemia/complications , Cardiomyopathies/diagnosis , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Angina Pectoris/drug therapy , Fibrosis , Inflammation/drug therapy
12.
J Biochem Mol Toxicol ; 37(7): e23366, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37063075

ABSTRACT

Myocardial ischemia-reperfusion injury (MI/R) leads to the inevitable clinical consequences of myocardial infarction and subsequent heart failure. Trimetazidine (TMZ), an anti-ischemic agent, exerts protective potential in MI/R but had limited efficacy for some patients. Here we sought to investigate the single and combined application of gentiopicroside (GPS) and TMZ in MI/R. Notably, GPS had little cytotoxicity to cardiomyocytes. GPS attenuated hypoxia/reoxygenation (H/R)-induced cell death, reactive oxygen species production, lactate dehydrogenase and malondialdehyde releases, and antioxidant stress enzyme superoxide dismutase activity, indicating the protective efficacy of GPS against H/R-induced oxidative injury. Importantly, GPS enhanced the protective efficacy of TMZ against H/R-mediated cardiomyocyte injury. Additionally, GPS mitigated the transcription and releases of pro-inflammatory cytokine interleukin-6 and tumor necrosis factor-α in H/R-treated cardiomyocytes, which were enhanced after co-treatment with TMZ. Mechanistically, GPS activated the AMP-activated protein kinase (AMPK) signaling to inhibit H/R-induced NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome, which was further enhanced after costimulation with TMZ. Importantly, blocking the AMPK signaling reversed the protective roles of GPS and its combination with TMZ in H/R-induced oxidative insult and inflammation. In vivo, both GPS and TMZ alleviated the abnormal cardiac structure, cardiomyocyte apoptosis, and cardiac dysfunction in MI/R rats, which were further enhanced after administration with GPS and TMZ together. Furthermore, GPS intensified TMZ-mediated inhibition of oxidative injury, inflammation, and the AMPK/NLRP3 signaling in MI/R rats. Collectively, GPS enhances the protective efficacy of TMZ against MI/R injury through AMPK activation-mediated inhibition of NLRP3 inflammasome signaling, implying a promising therapeutic agent for the treatment of MI/R.


Subject(s)
Myocardial Reperfusion Injury , Trimetazidine , Rats , Animals , Trimetazidine/pharmacology , Myocardial Reperfusion Injury/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , AMP-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Inflammation/metabolism
13.
Drug Chem Toxicol ; : 1-8, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37528808

ABSTRACT

Trimetazidine (TMZ) is a promising emerging therapeutic piperazine derivative for renal pathologies. However, the nephroprotective mechanism of TMZ against heavy metal-induced toxicity is unknown. This study, therefore, aimed to explore whether TMZ could mitigate mercury-induced nephrotoxicity in rats. Rats were injected TMZ (3 mg/kg bw) and/or mercury chloride (HgCl2) (4 mg/kg bw) for 4 days (n = 6 rats per group). The blood analysis revealed marked increases in creatinine, urea and uric acid levels in HgCl2 group compared to the control. HgCl2 induced prominent decreases in renal superoxide dismutase (SOD), catalase (CAT), glutathione peroxide (GPx) activities compared to the control followed by marked increases in the levels of malondialdehyde (MDA), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), caspase-3 and caspase-9. Whereas the renal levels of anti-inflammatory cytokines interleukin-4 (IL-4) and interleukin-10 (IL-10) reduced considerably compared to the control. Contrarily, it was found that in the rats administered TMZ + HgCl2, levels of renal markers, MDA, TNF-α, IL-6 and caspases-3/-9 were prominently reduced compared to the HgCl2 group. The renal SOD, CAT, GPx, IL-4, and IL-10 were markedly elevated along with ameliorated histopathological lesions. On the whole, therefore, TMZ could be repurposed for blocking HgCl2 nephrotoxicity via inhibition of oxidative inflammation and apoptosis in rats.

14.
Toxicol Mech Methods ; 33(7): 607-623, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37051630

ABSTRACT

Tramadol (TRA) causes neurotoxicity whereas trimetazidine (TMZ) is neuroprotective. The potential involvement of the PI3K/Akt/mTOR signaling pathway in the neuroprotection of TMZ against TRA-induced neurotoxicity was evaluated. Seventy male Wistar rats were divided into groups. Groups 1 and 2 received saline or TRA (50 mg/kg). Groups 3, 4, and 5 received TRA (50 mg/kg) and TMZ (40, 80, or 160 mg/kg) for 14 days. Group 6 received TMZ (160 mg/kg). Hippocampal neurodegenerative, mitochondrial quadruple complex enzymes, phosphatidylinositol-3-kinases (PI3Ks)/protein kinase B levels, oxidative stress, inflammatory, apoptosis, autophagy, and histopathology were evaluated. TMZ decreased anxiety and depressive-like behavior induced by TRA. TMZ in tramadol-treated animals inhibited lipid peroxidation, GSSG, TNF-α, and IL-1ß while increasing GSH, SOD, GPx, GR, and mitochondrial quadruple complex enzymes in the hippocampus. TRA inhibited Glial fibrillary acidic protein expression and increased pyruvate dehydrogenase levels. TMZ reduced these changes. TRA decreased the level of JNK and increased Beclin-1 and Bax. TMZ decreased phosphorylated Bcl-2 while increasing the unphosphorylated form in tramadol-treated rats. TMZ activated phosphorylated PI3Ks, Akt, and mTOR proteins. TMZ inhibited tramadol-induced neurotoxicity by modulating the PI3K/Akt/mTOR signaling pathways and its downstream inflammatory, apoptosis, and autophagy-related cascades.


Subject(s)
Neuroprotective Agents , Neurotoxicity Syndromes , Tramadol , Trimetazidine , Male , Rats , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Trimetazidine/pharmacology , Tramadol/toxicity , Neuroprotection , Rats, Wistar , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Apoptosis , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Autophagy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
15.
Ann Pharm Fr ; 81(1): 64-73, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35803334

ABSTRACT

OBJECTIVE: A simple and robust head space/gas chromatography with flame ionisation sensor (HS/GC/FIS) approach for the trace evaluation of carcinogenic impurity, methyl chloride, in trimetazidine dihydrochloride (TRD) drug ingredient and its formulation is described. METHOD: This HS/GC/FIS approach was based on separation and analysis of CH3Cl content on DB-624 [75.0m - length, 0.53mm - internal diameter, 3.0µm - film thickness] column using nitrogen as carrier gas flowing through the column at 3mL/min stream rate. Detection of eluted CH3Cl was accomplished with flame ionization sensor at a set temperature of 260̊C. RESULTS: The optimised HS/GC/FIS methodological approach was thoroughly validated, demonstrating that it was linear with range of 5.0ppm to 1508.4ppm, sensitive with detection limit of 1.65ppm and quantification limit of 5.01ppm, reproducible with RSD values of 2.10-2.35%, accurate with recoveries of 81.9-99.0%, robust with percent variation of 7.5-12.22% with respect to changes in oven temperature, injector temperature, detector temperature and practical for regular TRD quality control. CONCLUSION: The findings revealed that with this optimised HS/GC/FIS methodological approach, the trace amounts of carcinogenic impurity (methyl chloride) in TRD drug ingredient and formulation could be successfully measured.


Subject(s)
Methyl Chloride , Trimetazidine , Trimetazidine/analysis , Methyl Chloride/analysis , Carcinogens/analysis , Chromatography, Gas/methods , Temperature , Flame Ionization
16.
Wiad Lek ; 76(7): 1621-1626, 2023.
Article in English | MEDLINE | ID: mdl-37622506

ABSTRACT

OBJECTIVE: The aim: To study the parameters of the left ventricular (LV) diastolic function in patients with HT with concomitant T2DM and without it before and after complex treatment with the inclusion of Eplerenone 50 mg per day and Trimetazidine 80 mg per day during 3 months. PATIENTS AND METHODS: Materials and methods: The study included 50 patients, aged 45-54 years (mean age 51.3«1.5 years), women - 24 and men 26 with HT stage II. All patients were divided into 2 groups: 1 group (n=25) - patients with HT stage II (HbA1c level of 5.01«0.13%) and 2 group (n=25) - patients with HT stage II and concomitant T2DM (HbA1c level of 7.6«0.34%). The control group consisted of 20 healthy individuals (HbA1c level of 4.68«0.49%). RESULTS: Results: When analyzing the findings on left atrial volume index (LAVI), the highest indicators were observed in patients with HT with T2DM, but slightly lower in HT, and even lower in the control group, but the differences at this stage were not significant. This suggests that functional changes in cardiomyocyte kinetics, which develop in patients with comorbid pathology and are caused by metabolic and hemodynamic disorders, can progress steadily. CONCLUSION: Conclusions: After a three-month course of treatment with Eplerenone and Trimetazidine, the rate of myocardial relaxation in diastole likely increased in both groups of those examined. The prescribed treatment with Eplerenone and Trimetazidine has led to a decrease in the rate of progression of heart failure and a reduction in cardiovascular risks.


Subject(s)
Diabetes Mellitus, Type 2 , Hypertension , Trimetazidine , Ventricular Dysfunction, Left , Male , Humans , Female , Middle Aged , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Eplerenone , Glycated Hemoglobin , Hypertension/complications , Hypertension/drug therapy , Ventricular Dysfunction, Left/complications , Ventricular Dysfunction, Left/drug therapy
17.
Inflamm Res ; 71(2): 227-241, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34993560

ABSTRACT

OBJECTIVE: Trimetazidine (TMZ) exerts a strong inhibitory effect on ischemia/reperfusion (I/R) injury. Inflammation plays a key role in I/R injury. We hypothesized that TMZ may protect cardiomyocytes from I/R injury by inhibiting inflammation. METHODS: The left anterior descending coronary artery was ligated for 30 min followed by 6 h of reperfusion to establish a model of I/R injury. H9c2 cardiomyocytes were subjected to 2 h of hypoxia and 3 h of normoxic conditions to establish a model of hypoxia/reoxygenation (H/R) injury. We monitored the change in pyroptosis by performing Western blot analysis, microscopy and ELISA. RESULTS: I/R and H/R treatment stimulated gasdermin D-N domain (GSDMD-N) expression in cardiomyocytes (sham onefold vs. I/R 2.5-fold; control onefold vs. H/R 2.0-fold). Moreover, TMZ increased the viability of H9c2 cardiomyocytes subjected to H/R treatment (H/R 65.0% vs. H/R + TMZ 85.3%) and reduced the infarct size in vivo (I/R 47.0% vs. I/R + TMZ 28.3%). H/R and I/R treatment increased the levels of TLR4, MyD88, phospho-NF-κB p65 and the NLRP3 inflammasome; however, TMZ reduced the expression of these proteins. Additionally, TMZ inhibited noncanonical inflammasome signaling induced by I/R injury. CONCLUSIONS: In summary, TMZ alleviated pyroptosis induced by myocardial I/R injury through the TLR4/MyD88/NF-κB/NLRP3 inflammasome pathway. Therefore, TMZ represents an alternative treatment for myocardial I/R injury.


Subject(s)
Myocardial Reperfusion Injury/drug therapy , Phosphate-Binding Proteins/antagonists & inhibitors , Pore Forming Cytotoxic Proteins/antagonists & inhibitors , Pyroptosis/drug effects , Trimetazidine/pharmacology , Animals , Male , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NF-kappa B/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Phosphate-Binding Proteins/physiology , Pore Forming Cytotoxic Proteins/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Toll-Like Receptor 4/physiology
18.
J Nucl Cardiol ; 29(5): 2497-2507, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34331217

ABSTRACT

BACKGROUND: 18F-FDG PET myocardial metabolic imaging (MMI) is sometimes uninterpretable due to background activity from uncontrolled glucose homeostasis in diabetic mellitus (DM) patients. Trimetazidine is an oral medication that promotes the transformation of myocardial energy supply from free fatty acids to glucose. We aimed to investigate the feasibility and application of trimetazidine in 18F-FDG PET MMI of DM patients. METHODS: With DM patients exhibiting severe coronary artery disease (CAD) symptoms serving as self-controls, the effects of trimetazidine on PET MMI image quality, myocardial viability assessment, quantitative analytical parameters, and 18F-FDG uptake of different myocardial segments were elucidated. RESULTS: The image quality of 18F-FDG MMI was graded visually as good, moderate, and uninterpretable. After trimetazidine, grades of good, moderate, and uninterpretable were observed in 14 (60.9%), 8 (34.8%), and 1 (4.3%) patients, respectively, and in 4 (17.4%), 15 (65.2%), 4 (17.4%) patients without trimetazidine. The myocardial SUV and myocardial to blood pool SUV ratio (M/B ratio) were significantly higher after trimetazidine administration than those before (3.11 ± 1.07 vs 2.32 ± 1.00, 2.67 ± 1.41 vs 1.81 ± 0.75, P all < 0.01). 6 (3, 7) viable myocardium segments were detected with a mismatch score of 10 (6, 17) after trimetazidine, significantly higher than those before trimetazidine [5 (2, 7) and 8 (2, 17), P < 0.05]. Meanwhile, the 18F-FDG uptake in myocardial segments with decreased and normal perfusion showed different ranges of increase (by 15.30%-57.77%). CONCLUSION: Trimetazidine is feasible and effective in DM patients with severe CAD before 18F-FDG PET MMI, which can significantly improve the image quality and increase the number of viable myocardium segments detected. TRIAL REGISTRY: The study was registered in the Chinese Clinical Trial Registry (ChiCTR2000038559).


Subject(s)
Coronary Artery Disease , Diabetes Mellitus , Myocardial Perfusion Imaging , Trimetazidine , Coronary Artery Disease/complications , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/metabolism , Fatty Acids, Nonesterified/metabolism , Feasibility Studies , Fluorodeoxyglucose F18/metabolism , Glucose/metabolism , Humans , Myocardium/metabolism , Prospective Studies , Radiopharmaceuticals , Tomography, X-Ray Computed , Trimetazidine/metabolism
19.
J Biochem Mol Toxicol ; 36(8): e23114, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35633067

ABSTRACT

Late diagnosis and treatment of ovarian ischemia can lead to worsening of ischemia, irreversible damage to ovarian functions and infertility. In this process, there is no approved medical treatment that can reduce the negative effects of ischemia and contribute positively to ovarian functions during reperfusion after detorsion. Rats were randomly assigned into one of six groups of eight animals each. The groups were designed as follows: The control group, The ischemia(I) group, The Ischemia + Trimetazidine (I + TMZ) (20 mg/kg) group, and The ischemia-reperfusion group (I/R). The Ischemia-Reperfusion + Trimetazidine (I/R + TMZ) (20 mg/kg) group, and The Sham + Trimetazidine (Sham + TMZ) (20 mg/kg) group. In this study performed thiobarbituric acid reactive substances (TBARS), total thiol (-SH), interleukin 1 beta (IL-1ß), interleukin 6 (IL-6), toll-like receptor 4 (TLR4), and nuclear factor-kappa B(NF-κß). Increased oxidative stress and inflammation were as a result of ovarian I and I/R application. Trimetazidine (TMZ), was sufficient to reduce the oxidative stress and inflammation. TLR4 and NF-κß, which were upregulated by oxidative stress and inflammation, were regressed by TMZ. TMZ should be considered as a potential therapeutic agent in addition to surgery in the clinical treatment of ovarian torsion.


Subject(s)
Reperfusion Injury , Trimetazidine , Animals , Female , Inflammation/drug therapy , Ischemia/drug therapy , NF-kappa B/metabolism , Ovary/metabolism , Rats , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Signal Transduction , Toll-Like Receptor 4 , Trimetazidine/pharmacology , Trimetazidine/therapeutic use
20.
J Biochem Mol Toxicol ; 36(7): e23071, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35403780

ABSTRACT

Pulmonary fibrosis has been recently linked to metabolic dysregulation. Silica-induced pulmonary fibrosis in rats was employed by the current study to explore the effects of trimetazidine (a metabolic modulator-antianginal drug; TMZ) on silica-induced pulmonary fibrosis. Pulmonary fibrosis was induced by intranasal instillation of silica (50 mg/100 µl/rat) in TMZ versus vehicle-treated rats. Body weights of rats, weights of lungs, and wet-to-dry lung weights were determined. Various parameters were also measured in serum, bronchoalveolar lavage fluid (BALF) in addition to lung tissue homogenates. Moreover, histopathological examination of sectioned lungs for lesion score and distribution and histochemical detection of myeloperoxidase (MPO) in lung tissues were also performed. No significant differences were observed in body weight gains, lung coefficients, lung weights, and wet-to-dry lung weight in silica versus control rats. Elevated lactate levels in serum and lung homogenates were significantly attenuated by TMZ. In addition, lactate dehydrogenase activity, transforming growth factor-ß, and total proteins in BALF were significantly normalized with TMZ. Moreover, TMZ significantly increased reduced glutathione and adenosine triphosphate levels and decreased nitrate/nitrite and hydroxyproline content in lungs of silica-treated rats. Histopathological examination of lungs revealed more than 56% reduction in lesion score and distribution by TMZ. MPO expression in lungs of silica-treated rats was also significantly attenuated by TMZ. TMZ attenuates silica-induced pulmonary fibrosis, an effect that could be mediated by suppressing anaerobic glycolysis-induced excessive lactate production. Regulation of oxidative stress could also play a role in TMZ-promoted protective effects.


Subject(s)
Protective Agents/pharmacology , Pulmonary Fibrosis , Trimetazidine , Animals , Bronchoalveolar Lavage Fluid/chemistry , Lactates/analysis , Lactates/metabolism , Lactates/pharmacology , Lung/metabolism , Protective Agents/therapeutic use , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Rats , Silicon Dioxide/analysis , Silicon Dioxide/toxicity , Trimetazidine/pharmacology , Trimetazidine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL